Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3669, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38693119

ABSTRACT

Oncolytic viruses (OVs) show promise as a cancer treatment by selectively replicating in tumor cells and promoting antitumor immunity. However, the current immunogenicity induced by OVs for tumor treatment is relatively weak, necessitating a thorough investigation of the mechanisms underlying its induction of antitumor immunity. Here, we show that HSV-1-based OVs (oHSVs) trigger ZBP1-mediated PANoptosis (a unique innate immune inflammatory cell death modality), resulting in augmented antitumor immune effects. Mechanistically, oHSV enhances the expression of interferon-stimulated genes, leading to the accumulation of endogenous Z-RNA and subsequent activation of ZBP1. To further enhance the antitumor potential of oHSV, we conduct a screening and identify Fusobacterium nucleatum outer membrane vesicle (Fn-OMV) that can increase the expression of PANoptosis execution proteins. The combination of Fn-OMV and oHSV demonstrates potent antitumor immunogenicity. Taken together, our study provides a deeper understanding of oHSV-induced antitumor immunity, and demonstrates a promising strategy that combines oHSV with Fn-OMV.


Subject(s)
Fusobacterium nucleatum , Herpesvirus 1, Human , Oncolytic Virotherapy , Oncolytic Viruses , RNA-Binding Proteins , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/genetics , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Animals , Humans , Oncolytic Virotherapy/methods , Mice , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/immunology , Cell Line, Tumor , Fusobacterium nucleatum/immunology , Neoplasms/therapy , Neoplasms/immunology , Female , Immunity, Innate , Mice, Inbred BALB C
2.
iScience ; 26(6): 106916, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37305703

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) has been demonstrated to play a critical role in regulating cholesterol homeostasis and T cell antitumor immunity. However, the expression, function, and therapeutic value of PCSK9 in head and neck squamous cell carcinoma (HNSCC) remain largely unexplored. Here, we found that the expression of PCSK9 was upregulated in HNSCC tissues, and higher PCSK9 expression indicated poorer prognosis in HNSCC patients. We further found that pharmacological inhibition or siRNA downregulating PCSK9 expression suppressed the stemness-like phenotype of cancer cells in an LDLR-dependent manner. Moreover, PCSK9 inhibition enhanced the infiltration of CD8+ T cells and reduced the myeloid-derived suppressor cells (MDSCs) in a 4MOSC1 syngeneic tumor-bearing mouse model, and it also enhanced the antitumor effect of anti-PD-1 immune checkpoint blockade (ICB) therapy. Together, these results indicated that PCSK9, a traditional hypercholesterolemia target, may be a novel biomarker and therapeutic target to enhance ICB therapy in HNSCC.

3.
Int Immunopharmacol ; 119: 110243, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37137265

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is implicated in promoting HNSCC malignant progression. However, EZH2 inhibitors, when used alone, increase the number of myeloid-derived suppressor cells (MDSCs), which are responsible for enhancing tumor stemness and promoting tumor immune escape. We aimed to determine whether combining tazemetostat (an EZH2 inhibitor) and sunitinib (a MDSC inhibitor) can improve the response rate to an immune-checkpoint-blocking (ICB) therapy. We evaluated the efficacy of the above treatment strategies by bioinformatics analysis and animal experiments. EZH2 overexpression and abundant MDSCs in patients with HNSCC are associated with tumor progression. Tazemetostat treatment alone had limited inhibitory effect on HNSCC progression in the mouse models, accompanied by a surge in the number of MDSCs in the tumor microenvironment. Conversely, the combined use of tazemetostat and sunitinib reduced the number of MDSCs and regulatory T cell populations, promoting intratumoral infiltration of T cells and inhibiting of T cell exhausting, regulating of wnt/ß-catenin signaling pathway and tumor stemness, promoting the intratumoral PD-L1 expression and improved the response rate to anti-PD-1 therapy. The combined use of EZH2 and MDSC inhibitors effectively reverses HNSCC-specific immunotherapeutic resistance and is a promising strategy for overcoming resistance to ICB therapy.


Subject(s)
Head and Neck Neoplasms , Myeloid-Derived Suppressor Cells , Mice , Animals , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Sunitinib/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Tumor Microenvironment
4.
Cancer Lett ; 563: 216193, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37088326

ABSTRACT

Immunotherapy, particularly immune checkpoint blockade (ICB), has shown great promise in the treatment of cancer and emerged as a beacon of hope for patients who have exhausted traditional therapeutic options. Despite ICB's approval for the treatment of advanced tumors, its efficacy remains limited to a small subset of patients. As a systemic disease, cancer can induce changes in the composition and function of the systemic immune system, and ICB resistance often involves a dialog between the tumor microenvironment (TME) and the systemic immune macroenvironment. While investigations into tumor progression and ICB resistance have largely focused on the TME itself, the alterations in the systemic immune system and immune macroenvironment are still poorly understood. Given the spleen's role as the largest secondary lymphoid organ, its examination and discussion may provide valuable insights into the systemic immune status and TME components. Recent studies have highlighted the importance of the spleen in tumor progression and immunotherapy, particularly in the context of erythroid progenitor cells (EPCs), a significant cell subpopulation. In this review, we discuss the mechanisms and role of splenic extramedullary hematopoiesis (EMH) as an intermediary in tumor-host interactions and explore the mechanism of EPC-TME collusions. We further summarize the progress in EPC-targeting strategies and emphasize the potential for further research into the role and mechanisms of EPCs in tumor progression and treatment, which could have far-reaching implications.


Subject(s)
Erythroid Precursor Cells , Neoplasms , Humans , Neoplasms/therapy , Immunotherapy , Spleen , Tumor Microenvironment
5.
Mol Ther ; 31(7): 2154-2168, 2023 07 05.
Article in English | MEDLINE | ID: mdl-36869589

ABSTRACT

Immune checkpoint blockade (ICB) treatment has demonstrated excellent medical effects in oncology, and it is one of the most sought after immunotherapies for tumors. However, there are several issues with ICB therapy, including low response rates and a lack of effective efficacy predictors. Gasdermin-mediated pyroptosis is a typical inflammatory death mode. We discovered that increased expression of gasdermin protein was linked to a favorable tumor immune microenvironment and prognosis in head and neck squamous cell carcinoma (HNSCC). We used the mouse HNSCC cell lines 4MOSC1 (responsive to CTLA-4 blockade) and 4MOSC2 (resistant to CTLA-4 blockade) orthotopic models and demonstrated that CTLA-4 blockade treatment induced gasdermin-mediated pyroptosis of tumor cells, and gasdermin expression positively correlated to the effectiveness of CTLA-4 blockade treatment. We found that CTLA-4 blockade activated CD8+ T cells and increased the levels of interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α) cytokines in the tumor microenvironment. These cytokines synergistically activated the STAT1/IRF1 axis to trigger tumor cell pyroptosis and the release of large amounts of inflammatory substances and chemokines. Collectively, our findings revealed that CTLA-4 blockade triggered tumor cells pyroptosis via the release of IFN-γ and TNF-α from activated CD8+ T cells, providing a new perspective of ICB.


Subject(s)
CD8-Positive T-Lymphocytes , Head and Neck Neoplasms , Mice , Animals , Squamous Cell Carcinoma of Head and Neck/therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , CTLA-4 Antigen , Tumor Necrosis Factor-alpha/metabolism , Pyroptosis , Gasdermins , Cytokines/metabolism , Interferon-gamma/metabolism , Head and Neck Neoplasms/metabolism , Tumor Microenvironment
6.
Br J Cancer ; 128(11): 2126-2139, 2023 06.
Article in English | MEDLINE | ID: mdl-36977825

ABSTRACT

BACKGROUND: Enhancing the response rate of immunotherapy will aid in the success of cancer treatment. Here, we aimed to explore the combined effect of immunogenic radiotherapy with anti-PD-L1 treatment in immunotherapy-resistant HNSCC mouse models. METHODS: The SCC7 and 4MOSC2 cell lines were irradiated in vitro. SCC7-bearing mice were treated with hypofractionated or single-dose radiotherapy followed by anti-PD-L1 therapy. The myeloid-derived suppressive cells (MDSCs) were depleted using an anti-Gr-1 antibody. Human samples were collected to evaluate the immune cell populations and ICD markers. RESULTS: Irradiation increased the release of immunogenic cell death (ICD) markers (calreticulin, HMGB1 and ATP) in SCC7 and 4MOSC2 in a dose-dependent manner. The supernatant from irradiated cells upregulated the expression of PD-L1 in MDSCs. Mice treated with hypofractionated but not single-dose radiotherapy were resistant to tumour rechallenge by triggering ICD, when combined with anti-PD-L1 treatment. The therapeutic efficacy of combination treatment partially relies on MDSCs. The high expression of ICD markers was associated with activation of adaptive immune responses and a positive prognosis in HNSCC patients. CONCLUSION: These results present a translatable method to substantially improve the antitumor immune response by combining PD-L1 blockade with immunogenic hypofractionated radiotherapy in HNSCC.


Subject(s)
Head and Neck Neoplasms , Immune Checkpoint Inhibitors , Myeloid-Derived Suppressor Cells , Squamous Cell Carcinoma of Head and Neck , Animals , Humans , Mice , B7-H1 Antigen/metabolism , Head and Neck Neoplasms/drug therapy , Immunotherapy/methods , Myeloid-Derived Suppressor Cells/metabolism , Prognosis , Squamous Cell Carcinoma of Head and Neck/drug therapy , Immune Checkpoint Inhibitors/therapeutic use
7.
Oral Oncol ; 138: 106331, 2023 03.
Article in English | MEDLINE | ID: mdl-36753904

ABSTRACT

OBJECTIVES: CD103+CD8+T cells is a subtype of T cells with excellent tumor killing ability and it could response to immune checkpoint blockade therapy in several types of cancer, but the phenotype, role and molecular mechanism CD103+CD8+T cells in the OSCC still unclear. MATERIALS AND METHODS: The distribution and phenotype of CD103+CD8+T cells were investigated by performing multiplexed immunohistochemistry on human OSCC tissue microarray and flow cytometric analysis of fresh OSCC tumor-infiltrating lymphocytes (TILs). By in vivo use of anti-CD103 monoclonal antibody (mAb) in the 4MOSC1 tumor-bearing mouse model, CD103+CD8+T cell infiltration and cytotoxicity was clarified. RESULTS: The majority of CD8+T cells in both human and animal OSCC intra-tumoral region were CD103+CD8+T cells with high expression levels of cytotoxic molecules, which can be impaired by CD103 blockade. In addition, combined use of anti-CD103 mAb with anti-CTLA-4 mAb displayed impaired immune checkpoint blockade therapy efficiency. CONCLUSION: CD103+CD8+T cells are the major intra-tumoral subset of CD8+T cells in both animal and human OSCC, and that CD103+CD8+T cells demonstrate remarkable tumor-infiltrating and tumor-killing properties, thereby CD103+CD8+T cells may critical for anti-CTLA-4 immunotherapy in OSCC.


Subject(s)
Immune Checkpoint Inhibitors , Mouth Neoplasms , Humans , Animals , Mice , Mouth Neoplasms/metabolism , CD8-Positive T-Lymphocytes , Phenotype , Immunotherapy , Lymphocytes, Tumor-Infiltrating
8.
Adv Healthc Mater ; 12(7): e2202135, 2023 03.
Article in English | MEDLINE | ID: mdl-36479643

ABSTRACT

Pyroptosis is demonstrated to trigger antitumor immunity and represents a promising new strategy to potentiate cancer immunotherapy. The number of potent pyroptosis inducers, however, is limited and without tumor-targeting capability, which inevitably causes damage in normal tissues. Herein, a small molecular prodrug of paclitaxel-oxaliplatin is rationally synthesized, which can be covalently self-assembled with diselenide-containing cross-linking (Dse11), producing a diselenide nanoprodrug (DSe@POC) to induce pyroptosis for the first time. The diselenide bonds within DSe@POC can be split by high glutathione in the tumor microenvironment (TME) and reactive oxygen species induced by photodynamic therapy, thus possessing excellent TME on-target effects. Additionally, DSe@POC is able to elicit intense pyroptosis to remodel the immunostimulated TME and trigger a robust immune response. Furthermore, combined αPD-1 therapy effectively inhibits the growth of remote tumors through the abscopal effect, amplifies a long-term immune memory response to reject rechallenged tumors, and prolongs survival. Collectively, DSe@POC, as the first TME dual-responsive diselenide-based pyroptosis inducer, will open up an attractive approach for cancer immunotherapy.


Subject(s)
Neoplasms , Prodrugs , Humans , Prodrugs/pharmacology , Prodrugs/chemistry , Pyroptosis , Paclitaxel/pharmacology , Immunotherapy , Neoplasms/drug therapy , Tumor Microenvironment
9.
Sci Adv ; 8(47): eade3431, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36417504

ABSTRACT

The catalytic asymmetric geminal bis-nucleophilic addition to nonreactive functional groups is a type of highly desirable yet challenging transformation in organic chemistry. Here, we report the first catalytic asymmetric reductive/deoxygenative alkynylation of secondary amides. The method is based on a multicatalysis strategy that merges iridium/copper relay catalysis with organocatalysis. A further combination with the palladium-catalyzed alkyne hydrogenation allows the one-pot enantioselective reductive alkylation of secondary amides. This versatile protocol allows the efficient synthesis of four types of α-branched chiral amines, which are prevalent structural motifs of active pharmaceutical ingredients. The protocol also features excellent enantioselectivity, chemoselectivity, and functional group tolerance to be compatible with more reactive functional groups such as ketone and aldehyde. The synthetic utility of the method was further demonstrated by the late-stage functionalization of two drug derivatives and the concise, first catalytic asymmetric approach to the κ-opioid antagonist aticaprant.

10.
Acta Biomater ; 154: 497-509, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36367476

ABSTRACT

As the prominent feature of the development and progression of head and neck squamous cell carcinoma (HNSCC) is immunosuppression, therapeutic strategies to restore antitumor immunity have shown promising prospects. The efficacy of chemotherapy, a mainstay in HNSCC treatment, is exemplified by cytotoxic effects as well as immunostimulation, whereas compensatory activation of prosurvival signals in tumor tissues may compromise its efficacy. Aberrant activation of Src is present in many human malignancies including HNSCC, and is implicated in chemotherapy resistance. In this regard, tumor-microenvironment-responsive prodrug nanomicelles (PDO NPs) are rationally designed to combine chemotherapy (oxaliplatin, OXA) and Src inhibitors (dasatinib, DAS) for HNSCC therapy. PDO NPs are constructed by chemically modifying small-molecule prodrugs (DAS-OXA) loaded in block copolymer iPDPA with pH-triggered transforming capability. PDO NPs can controllably release drugs in response to tumor acidity, thus increasing tumor accumulation and therapeutic efficacy. Moreover, PDO NPs can elicit pyroptosis of tumor cells and induce T-cell-mediated antitumor immunity in murine HNSCC models. In summary, nanoprodrugs integrating Src inhibitors enhance the immunological effects of chemotherapy and provide insight into promising approaches for augmenting immunochemotherapy for HNSCC. STATEMENT OF SIGNIFICANCE: In this study, pH-responsive nanomicelles (PDO NPs) were constructed by loading a small molecular prodrug synthesized by the Src inhibitor dasatinib and the chemotherapy drug oxaliplatin into the amphiphilic block copolymer iPDPA to improve the immunological effects of chemotherapy for HNSCC. These nanomicelles can efficiently accumulate in tumor cells and achieve pH-responsive drug release. The PDO NPs can induce pyroptosis of tumor cells and potentiate antitumor immunity in subcutaneous and syngenetic orthotopic HNSCC mouse models, which may present a promising strategy to enhance immunochemotherapy for HNSCC.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Prodrugs , Mice , Humans , Animals , Head and Neck Neoplasms/drug therapy , Squamous Cell Carcinoma of Head and Neck/drug therapy , Dasatinib/pharmacology , Dasatinib/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Oxaliplatin , Prodrugs/pharmacology , Prodrugs/therapeutic use , Hydrogen-Ion Concentration , Cell Line, Tumor , Tumor Microenvironment
11.
Oral Dis ; 28(2): 364-372, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33386685

ABSTRACT

OBJECTIVES: Receptor for hyaluronic acid (HA)-mediated motility (RHAMM) is also known as CD168. This study proposed to elucidate the prognostic and clinicopathological significance of CD168 expression in oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS: Immune staining of a human tissue microarray and Western blot were used to reveal the expression level of CD168 in OSCC. Correlations between clinicopathological indexes and CD168 expression in OSCC patients were assessed. RESULTS: Increased expression of CD168 was detected in OSCC tissues. High expression of CD168 indicated worse survival of patients (p < .05). Furthermore, high expression of CD168 was related to pathological grade in OSCC (p < .05). CD168 expression was positively related to programmed death ligand 1 (PD-L1), CKLF-like MARVEL transmembrane domain-containing protein 6 (CMTM6), B7 homology 4 protein (B7-H4), CD44, CD133, and Slug expression in OSCC. CONCLUSION: This study revealed the overexpression of CD168 in OSCC and shed light on the prognostic significance of CD168 expression in OSCC patients.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Humans , Mouth Neoplasms/pathology , Prognosis , Squamous Cell Carcinoma of Head and Neck
12.
Oral Dis ; 27(2): 204-214, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32640108

ABSTRACT

OBJECTIVES: Ribonucleotide reductase M2 (RRM2) is a rate-limiting enzyme involved in DNA repair and synthesis. This study aimed to investigate the expression level, clinicopathological significance, and prognostic value of RRM2 in oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS: Human OSCC tissue microarrays were used to detect the expression of RRM2, cancer stem cell (CSC) markers CD44 and aldehyde dehydrogenase 1 (ALDH1), and the epithelial-mesenchymal transition (EMT) marker Slug. The correlation of RRM2 expression with clinicopathological parameters was evaluated. The effects of RRM2 on cell proliferation, migration, and apoptosis were investigated. RESULTS: Compared with normal and dysplastic tissues, the expression of RRM2 in human primary OSCC was significantly increased, and its overexpression was correlated with advanced pathological grade. The overall survival rate of patients with high RRM2 expression was lower than that of patients with low RRM2 expression. The overexpression of RRM2 was significantly associated with OSCC recurrence, and its overexpression was correlated with the CSC markers CD44 and ALDH1 and the EMT marker Slug. The expression of RRM2 promotes the proliferation and migration of human OSCC cells and inhibits apoptosis. CONCLUSION: Ribonucleotide reductase M2 may be a novel target in the diagnosis, prognosis, and therapy of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Carcinoma, Squamous Cell/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Mouth Neoplasms/genetics , Neoplasm Recurrence, Local , Prognosis , Ribonucleoside Diphosphate Reductase , Squamous Cell Carcinoma of Head and Neck
13.
Front Oncol ; 10: 588530, 2020.
Article in English | MEDLINE | ID: mdl-33163414

ABSTRACT

This study focused on the expression of mitochondrial serine hydroxymethyltransferase (SHMT2) in oral squamous cell carcinoma (OSCC) and its correlation with clinical traits and the prognosis of OSCC patients. Immunochemical staining and Western blotting were used to quantify the expression of SHMT2 and related immune markers in OSCC. Using OSCC microarrays and The Cancer Genome Atlas (TCGA) database, we evaluated the association between SHMT2 and various clinical traits. We found that increased expression of SHMT2 was detected in OSCC and correlated with advanced pathological grade and recurrence of OSCC. By a multivariate Cox proportional hazard model, high expression of SHMT2 was shown to indicate a negative prognosis. In addition, in the OSCC microenvironment, increasing the expression of SHMT2 was associated with high expression levels of programmed cell death-ligand 1 (PD-L1), CKLF-like MARVEL transmembrane domain containing 6 (CMTM6), V-type immunoglobulin domain-containing suppressor (VISTA), B7-H4, Slug, and CD317. In the future, more effort will be required to investigate the role of SHMT2 in the OSCC microenvironment.

14.
Int J Med Sci ; 17(11): 1598-1609, 2020.
Article in English | MEDLINE | ID: mdl-32669963

ABSTRACT

ATPase family AAA domain-containing protein 2 (ATAD2) is highly expressed in a variety of malignancies and can promote the proliferation of tumor cells and inhibit their differentiation. However, the expression of ATAD2 and its related mechanism in oral squamous cell carcinoma (OSCC) are still unknown. Immunohistochemical staining of ATAD2, cancer stem cells (CSCs) markers and immune checkpoint molecules was conducted on human OSCC specimens to determine the expression levels of these proteins and their correlations with the clinicopathological characteristics of ATAD2 in OSCC. Moreover, the role of ATAD2 in cell proliferation, apoptosis, migration and epithelial-mesenchymal transition (EMT) were assessed by silencing ATAD2 in vitro. Immunohistochemical analysis revealed that ATAD2 expression in OSCC tissues was markedly higher than that in adjacent dysplastic tissues and normal mucosal tissues. Overexpression of ATAD2 was related to poor overall survival in OSCC patients. In addition, the protein expression of ATAD2 was notably correlated with the expression of B7-H4, PD-L1, CMTM6, Slug and ALDH1 in human OSCC. ATAD2 knockdown arrested the cell cycle, promoted the apoptosis, and inhibited the proliferation, migration, and EMT of OSCC cells. In conclusion, these findings revealed that ATAD2 is highly expressed in OSCC and can act as a poor prognostic indicator.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , ATPases Associated with Diverse Cellular Activities/genetics , Apoptosis/genetics , Apoptosis/physiology , Carcinoma, Squamous Cell/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Mouth Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis
15.
Zhonghua Nan Ke Xue ; 24(11): 967-973, 2018 Nov.
Article in Chinese | MEDLINE | ID: mdl-32212468

ABSTRACT

OBJECTIVE: To establish a method for in vitro culture of the fetal rat testis tissue. METHODS: Nine sexually mature specific-pathogen-free rats, 3 males and 6 females, weighing 200-250 g, were used for this study. The estrus of the female rats was determined according to the results of the vaginal smear test. The female rats were mated with the male ones in proestrus and estrus at night in the ratio of 2∶1 and observed the following day for conception (0.5 day post-conception [dpc]) based on the presence of sperm in the vaginal smear. At 15.5 dpc, the fetal testes were isolated under the anatomical microscope, some for HE staining and the rest divided into a control and an hCG group to be cultured in a soft agar culture system at 37 ℃ in a humidified atmosphere containing 5% CO2. From the first day of culture (d 0), the development of the testes was observed under the inverted microscope, the culture medium collected and replaced on d 1, d 2, d 3 and d 4, and the testis tissue obtained on d 4. The concentration of testosterone in the culture medium was determined and the testis tissues were fixed, dehydrated and embedded for histological examination. RESULTS: Fetal rats were successfully obtained with the vaginal smear at 15.5 dpc, and the fetal testes effectively isolated, which were well developed, with gradual increase of their volume and enlargement of convoluted seminiferous tubules under the inverted microscope. Testosterone was observed in the culture medium, its concentration gradually increasing and reaching the peak on d 3, and its secretion stimulated by hCG. At 15.5 dpc. The fetal testes showed a histomorphological integrity, with typical seminiferous tubules, gonocytes, Sertoli cells and Leydig cells, but no central necrosis. Transmission electron microscopy revealed gonocytes and Sertoli cells within and Leydig cells between the seminiferous tubules, without obvious swelling of the mitochondria and endoplasmic reticula in the cells. CONCLUSIONS: The fetal rat testis tissue cultured in the soft agar culture system can develop well, retain its normal activity, and excrete testosterone into the culture medium.


Subject(s)
Cell Culture Techniques , Leydig Cells , Testis , Animals , Cell Culture Techniques/methods , Female , Male , Rats , Seminiferous Tubules , Sertoli Cells , Spermatozoa , Testis/cytology , Testosterone
16.
Neural Regen Res ; 11(12): 1981-1989, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28197196

ABSTRACT

Several studies have investigated the protective functions of brain-derived neurotrophic factor (BDNF) in retinitis pigmentosa. However, a BDNF-based therapy for retinitis pigmentosa is not yet available. To develop an efficient treatment for fundus disease, an eukaryotic expression plasmid was generated and used to transfect human 293T cells to assess the expression and bioactivity of BDNF on acute retinal pigment epithelial-19 (ARPE-19) cells, a human retinal epithelial cell line. After 96 hours of co-culture in a Transwell chamber, ARPE-19 cells exposed to BDNF secreted by 293T cells were more viable than ARPE-19 cells not exposed to secreted BDNF. Western blot assay showed that Bax levels were downregulated and that Bcl-2 levels were upregulated in human ARPE-19 cells exposed to BDNF. Furthermore, 293T cells transfected with the BDNF gene steadily secreted the protein. The powerful anti-apoptotic function of this BDNF may be useful for the treatment of retinitis pigmentosa and other retinal degenerative diseases.

SELECTION OF CITATIONS
SEARCH DETAIL
...